Elsevier

Life Sciences

Volume 88, Issues 17–18, 25 April 2011, Pages 737-745
Life Sciences

Minireview
Current issues with acetaminophen hepatotoxicity—A clinically relevant model to test the efficacy of natural products

https://doi.org/10.1016/j.lfs.2011.01.025Get rights and content

Abstract

There is a significant need to evaluate the therapeutic potential of natural products and other compounds purported to be hepatoprotective. Acetaminophen-induced liver injury, especially in mice, is an attractive and widely used model for this purpose because it is both clinically relevant and experimentally convenient. However, the pathophysiology of liver injury after acetaminophen overdose is complex. This review describes the multiple steps and signaling pathways involved in acetaminophen-mediated cell death. The toxicity is initiated by the formation of a reactive metabolite, which depletes glutathione and binds to cellular proteins, especially in mitochondria. The resulting mitochondrial oxidant stress and peroxynitrite formation, in part through amplification by c-jun-N-terminal kinase activation, leads to mitochondrial DNA damage and opening of the mitochondrial permeability transition pore. Endonucleases from the mitochondrial intermembrane space and lysosomes are responsible for nuclear DNA fragmentation. Despite the oxidant stress, lipid peroxidation is not a relevant mechanism of injury. The mitochondrial dysfunction and nuclear DNA damage ultimately cause oncotic necrotic cell death with release of damage-associated molecular patterns that trigger a sterile inflammatory response. Current evidence supports the hypothesis that innate immune cells do not contribute to injury but are involved in cell debris removal and regeneration. This review discusses the latest mechanistic aspects of acetaminophen hepatotoxicity and demonstrates ways to assess the mechanisms of drug action and design experiments needed to avoid pitfalls and incorrect conclusions. This review should assist investigators in the optimal use of this model to test the efficacy of natural compounds and obtain reliable mechanistic information.

Introduction

The search for new drugs and novel therapeutic intervention strategies increasingly includes testing plant extracts and other natural products. In addition, more products of traditional Eastern medicine are being evaluated. Independent of whether extracts are considered or if individual ingredients of a mixture are tested, the pharmacological efficacy of these chemicals needs to be investigated. For compounds that are assumed to have hepatoprotective effects, the model of acetaminophen (APAP) overdose in rodents, especially mice, is one of the most popular experimental in vivo systems used today (Campos et al., 1989, Chen et al., 2009, Gao and Zhou, 2005, Hau et al., 2009, Hsu et al., 2008, Küpeli et al., 2006, Wang et al., 2010, Wu et al., 2008, Wu et al., 2010, Yuan et al., 2010). The advantage of this model is that APAP is a dose-dependent hepatotoxicant, the experiments are technically easy to perform and, most importantly, it is a clinically relevant model. However, after more than 35 years of research there is substantial information in the literature on mechanisms of APAP hepatotoxicity (Hinson et al., 2004, Jaeschke et al., 2003, Jaeschke and Bajt, 2006, Jaeschke and Bajt, 2010, Nelson, 1990, Nelson and Bruschi, 2001). Some of these mechanisms are well established and others are more or less controversial; some are correct and some are skewed by experimental conditions making them not clinically applicable. In addition, many aspects of APAP-induced cell death and liver injury are still unknown. Thus, the mechanisms of APAP hepatotoxicity are extremely complex and the interpretation of in vivo data is difficult. Unfortunately, the model is being used as a tool by investigators who are not necessarily experts in APAP toxicity leading to frequent misinterpretation of data (Jaeschke et al., 2010). Therefore, the purpose of this review on APAP hepatotoxicity is to discuss some of the established and controversial mechanisms and the potential pitfalls one should be aware of when using this model.

Section snippets

Models of acetaminophen-induced liver injury

To study mechanisms of APAP toxicity, the mouse in vivo or primary mouse hepatocytes are most frequently used. Various strains of mice (outbred or inbred strains) are susceptible to APAP toxicity but some strain differences exist (Harrill et al., 2009). Mice are fasted overnight to reduce hepatic glutathione levels and are treated i.p. with doses of 200–400 mg/kg APAP dissolved in warm saline. Liver injury develops between 3 and 5 h and peaks at 12 h after APAP administration. Fasting allows lower

Metabolic activation of acetaminophen

One of the earliest works on mechanisms of APAP hepatotoxicity demonstrated that a small fraction of the dose is metabolized by the cytochrome P450 (cyp) system to form a reactive metabolite (Jollow et al., 1973, Mitchell et al., 1973a, Mitchell et al., 1973b). The metabolite was identified as N-acetyl-p-benzoquinone imine (NAPQI) (Dahlin et al., 1984). The primary enzyme involved is Cyp2E1, but others appear to have a role (Thummel et al., 1993, Wolf et al., 2007, Zaher et al., 1998).

Protein adducts of acetaminophen and oxidant stress

The early “protein binding” hypothesis was repeatedly questioned because the overall fraction of the administered dose that ends up covalently bound to cellular proteins is small and certain interventions appear to be able to separate protein binding from cell injury (Jørgensen et al., 1988). As a result, a competing hypothesis was introduced. Wendel and coworkers hypothesized that reactive oxygen generated during the metabolism of APAP can cause lipid peroxidation (LPO), which may be the

Sources of reactive oxygen and reactive nitrogen species

APAP overdose causes an oxidant stress (Jaeschke, 1990) and peroxynitrite formation (Hinson et al., 1998). The increase in tissue glutathione disulfide (GSSG) levels in vivo, as a specific marker for hydrogen peroxide, is caused by a selective accumulation of GSSG in mitochondria (Jaeschke, 1990, Knight et al., 2001). This suggests that enhanced amounts of superoxide are being generated by the electron transport chain and released into the mitochondrial matrix (Fig. 3). This conclusion is

Lipid peroxidation

Due to the involvement of oxidant stress, LPO is a popular hypothesis to explain massive cell death after APAP overdose. Antioxidant function and protection against LPO are probably the most invoked mechanisms of protection by natural products (Campos et al., 1989, Gao and Zhou, 2005, Hsu et al., 2008, Küpeli et al., 2006, Wang et al., 2010, Wu et al., 2008, Wu et al., 2010, Yuan et al., 2010). LPO is a multistep process requiring initiation of a radical chain reaction and propagation through

Necrosis and apoptosis

APAP-induced liver injury is characterized by extensive cell contents release (liver enzymes), cell swelling, nuclear degradation (karyorrhexis and karyolysis) and an inflammatory response (Gujral et al., 2002). These are typical characteristics of oncotic necrosis (Jaeschke and Lemasters, 2003). Thus, it is generally concluded that APAP-induced cell death in vivo (Gujral et al., 2002) and in vitro (Bajt et al., 2004, Kon et al., 2004) is caused by oncotic necrosis (Fig. 4). However, cell death

Innate immune response

The cell contents released after APAP-induced necrotic cell death initiates an inflammatory response with activation of Kupffer cells and recruitment of neutrophils and monocytes into the liver (Laskin, 2009, Laskin and Pilaro, 1986, Lawson et al., 2000) (Fig. 5). It was recently recognized that some of the compounds generally released by dying hepatocytes can stimulate toll-like receptors on macrophages and other non-parenchymal cells and promote cytokine formation initiating an inflammatory

Conclusions

APAP-induced liver injury, especially in mice, is a clinically relevant model that is suitable to test the efficacy of hepatoprotective natural products and other compounds in vivo. Given the extensive knowledge of the mechanisms of APAP-induced liver injury, the model can also be used to investigate mechanisms of therapeutic action. Importantly, though many studies demonstrate an antioxidant effect or protection against LPO with natural products after APAP treatment, this is highly unlikely

Conflict of interest statement

The authors declare that there are no conflicts of interest.

Acknowledgments

Research in the authors' laboratory is supported in part by National Institutes of Health Grants R01 DK070195 and R01 AA12916 (to H.J.) and by grants P20 RR016475 and P20 RR021940 from the National Center for Research Resources (NCRR), a component of the National Institutes of Health. C.D. Williams was supported by the “Training Program in Environmental Toxicology” (T32 ES007079-26A2) from the National Institute of Environmental Health Sciences.

References (113)

  • D.K. Hau et al.

    Phyllanthus urinaria extract attenuates acetaminophen induced hepatotoxicity: involvement of cytochrome P450 CYP2E1

    Phytomedicine

    (2009)
  • C.C. Hsu et al.

    Preventive effect of Ganoderma amboinense on acetaminophen-induced acute liver injury

    Phytomedicine

    (2008)
  • H. Jaeschke

    Antioxidant defense mechanisms

  • H. Jaeschke et al.

    Mechanisms of acetaminophen hepatotoxicity

  • H. Jaeschke et al.

    Apoptosis versus oncotic necrosis in hepatic ischemia/reperfusion injury

    Gastroenterology

    (2003)
  • H. Jaeschke et al.

    NADH-dependent reductive stress and ferritin-bound iron in allyl alcohol-induced lipid peroxidation in vivo: the protective effect of vitamin E

    Chem Biol Interact

    (1992)
  • H. Jaeschke et al.

    Protection against TNF-induced liver parenchymal cell apoptosis during endotoxemia by a novel caspase inhibitor in mice

    Toxicol Appl Pharmacol

    (2000)
  • H. Jaeschke et al.

    The role of oxidant stress and reactive nitrogen species in acetaminophen hepatotoxicity

    Toxicol Lett

    (2003)
  • H. Jaeschke et al.

    Role of caspases in acetaminophen-induced liver injury

    Life Sci

    (2006)
  • T.R. Knight et al.

    Acetaminophen-induced inhibition of Fas receptor-mediated liver cell apoptosis: mitochondrial dysfunction versus glutathione depletion

    Toxicol Appl Pharmacol

    (2002)
  • E. Küpeli et al.

    Effect of Cistus laurifolius L. leaf extracts and flavonoids on acetaminophen-induced hepatotoxicity in mice

    J Ethnopharmacol

    (2006)
  • D.L. Laskin et al.

    Potential role of activated macrophages in acetaminophen hepatotoxicity. I. Isolation and characterization of activated macrophages from rat liver

    Toxicol Appl Pharmacol

    (1986)
  • J.A. Lawson et al.

    Inhibition of Fas receptor (CD95)-induced hepatic caspase activation and apoptosis by acetaminophen in mice

    Toxicol Appl Pharmacol

    (1999)
  • Z.X. Liu et al.

    Innate immune system plays a critical role in determining the progression and severity of acetaminophen hepatotoxicity

    Gastroenterology

    (2004)
  • B.V. Martin-Murphy et al.

    The role of damage associated molecular pattern molecules in acetaminophen-induced liver injury in mice

    Toxicol Lett

    (2010)
  • Y. Masubuchi et al.

    Involvement of mitochondrial permeability transition in acetaminophen-induced liver injury in mice

    J Hepatol

    (2005)
  • W.R. Mathews et al.

    Lipid peroxidation as molecular mechanism of liver cell injury during reperfusion after ischemia

    Free Radic Biol Med

    (1994)
  • Y. Park et al.

    Prevention of acetaminophen-induced hepatotoxicity by dimethyl sulfoxide

    Toxicology

    (1988)
  • Y. Qiu et al.

    Identification of the hepatic protein targets of reactive metabolites of acetaminophen in vivo in mice using two-dimensional gel electrophoresis and mass spectrometry

    J Biol Chem

    (1998)
  • S.D. Ray et al.

    Early loss of large genomic DNA in vivo with accumulation of Ca2+ in the nucleus during acetaminophen-induced liver injury

    Toxicol Appl Pharmacol

    (1990)
  • C. Saito et al.

    c-Jun N-terminal kinase modulates oxidant stress and peroxynitrite formation independent of inducible nitric oxide synthase in acetaminophen hepatotoxicity

    Toxicol Appl Pharmacol

    (2010)
  • C. Saito et al.

    Mechanism of protection by metallothionein against acetaminophen hepatotoxicity

    Toxicol Appl Pharmacol

    (2010)
  • W. Shen et al.

    Acetaminophen-induced cytotoxicity in cultured mouse hepatocytes: correlation of nuclear Ca2+ accumulation and early DNA fragmentation with cell death

    Toxicol Appl Pharmacol

    (1991)
  • K.E. Thummel et al.

    Oxidation of acetaminophen to N-acetyl-p-aminobenzoquinone imine by human CYP3A4

    Biochem Pharmacol

    (1993)
  • M.A. Tirmenstein et al.

    Subcellular binding and effects on calcium homeostasis produced by acetaminophen and a nonhepatotoxic regioisomer, 3'-hydroxyacetanilide, in mouse liver

    J Biol Chem

    (1989)
  • M.L. Adams et al.

    Enhanced acetaminophen hepatotoxicity in transgenic mice overexpressing BCL-2

    Mol Pharmacol

    (2001)
  • D.J. Antoine et al.

    High-mobility group box-1 protein and keratin-18, circulating serum proteins informative of acetaminophen-induced necrosis and apoptosis in vivo

    Toxicol Sci

    (2009)
  • M.L. Bajt et al.

    Protection against Fas receptor-mediated apoptosis in hepatocytes and nonparenchymal cells by a caspase-8 inhibitor in vivo: evidence for a postmitochondrial processing of caspase-8

    Toxicol Sci

    (2000)
  • M.L. Bajt et al.

    Scavenging peroxynitrite with glutathione promotes regeneration and enhances survival during acetaminophen-induced liver injury in mice

    J Pharmacol Exp Ther

    (2003)
  • M.L. Bajt et al.

    Acetaminophen-induced oxidant stress and cell injury in cultured mouse hepatocytes: protection by N-acetyl cysteine

    Toxicol Sci

    (2004)
  • M.L. Bajt et al.

    Nuclear translocation of endonuclease G and apoptosis-inducing factor during acetaminophen-induced liver cell injury

    Toxicol Sci

    (2006)
  • M.L. Bajt et al.

    Mitochondrial bax translocation accelerates DNA fragmentation and cell necrosis in a murine model of acetaminophen hepatotoxicity

    J Pharmacol Exp Ther

    (2008)
  • M.E. Bianchi

    DAMPs, PAMPs and alarmins: all we need to know about danger

    J Leukoc Biol

    (2007)
  • M. Bilzer et al.

    Prevention of Kupffer cell-induced oxidant injury in rat liver by atrial natriuretic peptide

    Am J Physiol

    (1999)
  • R. Campos et al.

    Silybin dihemisuccinate protects against glutathione depletion and lipid peroxidation induced by acetaminophen on rat liver

    Planta Med

    (1989)
  • X. Chen et al.

    Protective effect of tea polyphenols against paracetamol-induced hepatotoxicity in mice is significantly correlated with cytochrome P450 suppression

    World J Gastroenterol

    (2009)
  • G.B. Corcoran et al.

    Effects of N-acetylcysteine on acetaminophen covalent binding and hepatic necrosis in mice

    J Pharmacol Exp Ther

    (1985)
  • C. Cover et al.

    Peroxynitrite-induced mitochondrial and endonuclease-mediated nuclear DNA damage in acetaminophen hepatotoxicity

    J Pharmacol Exp Ther

    (2005)
  • D.C. Dahlin et al.

    N-acetyl-p-benzoquinone imine: a cytochrome P-450-mediated oxidation product of acetaminophen

    Proc Natl Acad Sci U S A

    (1984)
  • H. Gao et al.

    Anti-lipid peroxidation and protection of liver mitochondria against injuries by picroside II

    World J Gastroenterol

    (2005)
  • Cited by (198)

    • Drug-induced oxidative stress as a mechanism of toxicity

      2023, Essentials of Pharmatoxicology in Drug Research: Toxicity and Toxicodynamics: Volume 1
    • Applicability of generic PBK modelling in chemical hazard assessment: A case study with IndusChemFate

      2022, Regulatory Toxicology and Pharmacology
      Citation Excerpt :

      The calculated equivalent external doses were analysed using the Benchmark Dose (BMD) approach, with the software PROAST (Slob 2002), so as to obtain the predicted in vivo dose-response curves. For a quantitative comparison with the usual range of acetaminophen human overdose, leading to hepatotoxicity (Bunchorntavakul and Reddy 2013; Jaeschke et al., 2011), 90% confidence intervals were estimated for the underlying BMD at 10% effect size (BMD10) and at 20% effect size (BMD20) (EFSA 2017). The confidence intervals are denoted by the lower (BMDL) and upper (BMDU) limits.

    View all citing articles on Scopus
    View full text